Skip to main content
Top
Published in: Respiratory Research 1/2015

Open Access 01-12-2015 | Research

Autophagy contributes to the chemo-resistance of non-small cell lung cancer in hypoxic conditions

Authors: Jin Gu Lee, Ju Hye Shin, Hyo Sup Shim, Chang Young Lee, Dae Joon Kim, Young Sam Kim, Kyung Young Chung

Published in: Respiratory Research | Issue 1/2015

Login to get access

Abstract

Background

The development of chemo-resistance in non-small lung cancer is a major obstacle in treating patients. Hypoxia is a commonly faced microenvironment in solid tumor and suggested to be related to both autophagy and chemo-resistance.

Methods

In this study, we investigated the role of hypoxia-induced autophagy in acquiring chemo-resistance in both cancer cell (A549) and human cancer tissue

Results

Hypoxic exposure (1 % O2) of A549 cell stimulated autophagic induction in cancer cells, shown by increase of LC3BI to LC3BII conversion and decrease of p62/sequestosome1 in Western blot, increased GFP-LC puncta in confocal microscopy, and increased number of double-membrane autophagic vacuoles in electron micrographs. Hypoxic exposure also induced resistance of cancer cells to cisplatin, and LC3B siRNA restored the sensitivity of cancer cells to chemotherapy. Furthermore, Human lung cancer tissues that experienced chemotherapy showed increase of LC3BI to LC3BII conversion and decrease of p62/sequestosome1 compared with chemo-naïve cancer tissue in Western blot.

Conclusion

Autophagy may play an important role in acquiring resistance to chemotherapy in lung cancer and hypoxia related pathway seems to be involved in autophagy induction.
Literature
1.
go back to reference Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J for clin. 2009;59:225–49.CrossRef Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J for clin. 2009;59:225–49.CrossRef
2.
go back to reference Chen N, Karantza-Wadsworth V. Role and regulation of autophagy in cancer. Biochim Biophys Acta. 1793;2009:1516–23. Chen N, Karantza-Wadsworth V. Role and regulation of autophagy in cancer. Biochim Biophys Acta. 1793;2009:1516–23.
4.
go back to reference Kondo Y, Kanzawa T, Sawaya R, Kondo S. The role of autophagy in cancer development and response to therapy. Nat Rev Cancer. 2005;5:726–34.CrossRefPubMed Kondo Y, Kanzawa T, Sawaya R, Kondo S. The role of autophagy in cancer development and response to therapy. Nat Rev Cancer. 2005;5:726–34.CrossRefPubMed
6.
go back to reference Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, et al. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell. 2005;120:237–48.CrossRefPubMed Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, et al. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell. 2005;120:237–48.CrossRefPubMed
7.
go back to reference Cosse JP, Michiels C. Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression. Anticancer Agents Med Chem. 2008;8:790–7.CrossRefPubMed Cosse JP, Michiels C. Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression. Anticancer Agents Med Chem. 2008;8:790–7.CrossRefPubMed
8.
go back to reference Zhou J, Schmid T, Schnitzer S, Brüne B. Tumor hypoxia and cancer progression. Cancer Lett. 2006;237:10–21.CrossRefPubMed Zhou J, Schmid T, Schnitzer S, Brüne B. Tumor hypoxia and cancer progression. Cancer Lett. 2006;237:10–21.CrossRefPubMed
9.
go back to reference Höckel M, Vaupel P. Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst. 2001;93:266–76.CrossRefPubMed Höckel M, Vaupel P. Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst. 2001;93:266–76.CrossRefPubMed
10.
go back to reference Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3:721–32.CrossRef Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3:721–32.CrossRef
11.
go back to reference Qie Y, Li P Ji C. Cell Death Conversion under Hypoxic Condition in Tumor Development and Therapy. Int JMil Sci. 2015;16(10):25536-51. Qie Y, Li P Ji C. Cell Death Conversion under Hypoxic Condition in Tumor Development and Therapy. Int JMil Sci. 2015;16(10):25536-51.
12.
go back to reference Rouschop KM, Wouters BG. Regulation of autophagy through multiple independent hypoxic signaling pathways. Curr Mol Med. 2009;9(4):417–24.CrossRefPubMed Rouschop KM, Wouters BG. Regulation of autophagy through multiple independent hypoxic signaling pathways. Curr Mol Med. 2009;9(4):417–24.CrossRefPubMed
13.
go back to reference Mazure NM, Pouysségur J. Hypoxia-induced autophagy: cell death or cell survival? Curr Opin Cell Biol. 2010;22(2):177–80.CrossRefPubMed Mazure NM, Pouysségur J. Hypoxia-induced autophagy: cell death or cell survival? Curr Opin Cell Biol. 2010;22(2):177–80.CrossRefPubMed
14.
go back to reference Lee CH, Lee MK, Kang CD, Kim YD, Park DY, Kim JY, et al. Differential expression of hypoxia inducible factor-1 alpha and tumor cell proliferation between squamous cell carcinomas and adenocarcinomas among operable non-small cell lung carcinomas. J Korean Med Sci. 2003;18:196–203.CrossRefPubMedPubMedCentral Lee CH, Lee MK, Kang CD, Kim YD, Park DY, Kim JY, et al. Differential expression of hypoxia inducible factor-1 alpha and tumor cell proliferation between squamous cell carcinomas and adenocarcinomas among operable non-small cell lung carcinomas. J Korean Med Sci. 2003;18:196–203.CrossRefPubMedPubMedCentral
15.
go back to reference Papandreou I, Lim A, Laderoute K, Denko N. Hypoxia signals autophagy in tumor cells via AMPK activity, independent of HIF-1, BNIP3, and BNIP3L. Cell Death Differ. 2008;15:1572–81.CrossRefPubMed Papandreou I, Lim A, Laderoute K, Denko N. Hypoxia signals autophagy in tumor cells via AMPK activity, independent of HIF-1, BNIP3, and BNIP3L. Cell Death Differ. 2008;15:1572–81.CrossRefPubMed
16.
go back to reference Abedin M, Wang D, McDonnell M, Lehmann U, Kelekar A. Autophagy delays apoptotic death in breast cancer cells following DNA damage. Cell Death Differ. 2006;14:500–10.CrossRef Abedin M, Wang D, McDonnell M, Lehmann U, Kelekar A. Autophagy delays apoptotic death in breast cancer cells following DNA damage. Cell Death Differ. 2006;14:500–10.CrossRef
17.
go back to reference Sato K, Tsuchihara K, Fujii S, Sugiyama M, Goya T, Atomi Y, et al. Autophagy is activated in colorectal cancer cells and contributes to the tolerance to nutrient deprivation. Cancer Res. 2007;67:9677.CrossRefPubMed Sato K, Tsuchihara K, Fujii S, Sugiyama M, Goya T, Atomi Y, et al. Autophagy is activated in colorectal cancer cells and contributes to the tolerance to nutrient deprivation. Cancer Res. 2007;67:9677.CrossRefPubMed
18.
go back to reference Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer cell. 2006;10:51–64.CrossRefPubMedPubMedCentral Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer cell. 2006;10:51–64.CrossRefPubMedPubMedCentral
19.
go back to reference Katayama M, Kawaguchi T, Berger M, Pieper R. DNA damaging agent-induced autophagy produces a cytoprotective adenosine triphosphate surge in malignant glioma cells. Cell Death Differ. 2006;14:548–58.CrossRefPubMed Katayama M, Kawaguchi T, Berger M, Pieper R. DNA damaging agent-induced autophagy produces a cytoprotective adenosine triphosphate surge in malignant glioma cells. Cell Death Differ. 2006;14:548–58.CrossRefPubMed
20.
21.
go back to reference Zhang H, Bosch-Marce M, Shimoda LA, Tan YS, Baek JH, Wesley JB, et al. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J Biol Chem. 2008;283:10892–903.CrossRefPubMedPubMedCentral Zhang H, Bosch-Marce M, Shimoda LA, Tan YS, Baek JH, Wesley JB, et al. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J Biol Chem. 2008;283:10892–903.CrossRefPubMedPubMedCentral
22.
go back to reference Pursiheimo J, Rantanen K, Heikkinen P, Johansen T, Jaakkola P. Hypoxia-activated autophagy accelerates degradation of SQSTM1/p62. Oncogene. 2008;28:334–44.CrossRefPubMed Pursiheimo J, Rantanen K, Heikkinen P, Johansen T, Jaakkola P. Hypoxia-activated autophagy accelerates degradation of SQSTM1/p62. Oncogene. 2008;28:334–44.CrossRefPubMed
23.
go back to reference Tracy K, Dibling BC, Spike BT, Knabb JR, Schumacker P, Macleod KF. BNIP3 is an RB/E2F target gene required for hypoxia-induced autophagy. Mol Cell Biol. 2007;27:6229–42.CrossRefPubMedPubMedCentral Tracy K, Dibling BC, Spike BT, Knabb JR, Schumacker P, Macleod KF. BNIP3 is an RB/E2F target gene required for hypoxia-induced autophagy. Mol Cell Biol. 2007;27:6229–42.CrossRefPubMedPubMedCentral
24.
go back to reference Schmaltz C, Hardenbergh PH, Wells A, Fisher DE. Regulation of proliferation-survival decisions during tumor cell hypoxia. Mol Cell Biol. 1998;18:2845–54.CrossRefPubMedPubMedCentral Schmaltz C, Hardenbergh PH, Wells A, Fisher DE. Regulation of proliferation-survival decisions during tumor cell hypoxia. Mol Cell Biol. 1998;18:2845–54.CrossRefPubMedPubMedCentral
25.
go back to reference Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol. 1992;12:5447–54.CrossRefPubMedPubMedCentral Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol. 1992;12:5447–54.CrossRefPubMedPubMedCentral
26.
go back to reference Hiraga T, Kizaka-Kondoh S, Hirota K, Hiraoka M, Yoneda T. Hypoxia and hypoxia-inducible factor-1 expression enhance osteolytic bone metastases of breast cancer. Cancer Res. 2007;67:4157.CrossRefPubMed Hiraga T, Kizaka-Kondoh S, Hirota K, Hiraoka M, Yoneda T. Hypoxia and hypoxia-inducible factor-1 expression enhance osteolytic bone metastases of breast cancer. Cancer Res. 2007;67:4157.CrossRefPubMed
27.
28.
go back to reference Smith TG, Robbins PA, Ratcliffe PJ. The human side of hypoxia‐inducible factor. Br J Haematol. 2008;141:325–34.CrossRefPubMed Smith TG, Robbins PA, Ratcliffe PJ. The human side of hypoxia‐inducible factor. Br J Haematol. 2008;141:325–34.CrossRefPubMed
29.
go back to reference Schnitzer S, Schmid T, Zhou J, Brüne B. Hypoxia and HIF-1α protect A549 cells from drug-induced apoptosis. Cell Death Differ. 2006;13:1611–3.CrossRefPubMed Schnitzer S, Schmid T, Zhou J, Brüne B. Hypoxia and HIF-1α protect A549 cells from drug-induced apoptosis. Cell Death Differ. 2006;13:1611–3.CrossRefPubMed
30.
go back to reference Pore N, Jiang Z, Gupta A, Cerniglia G, Kao GD, Maity A. EGFR tyrosine kinase inhibitors decrease VEGF expression by both hypoxia-inducible factor (HIF)-1–independent and HIF-1–dependent mechanisms. Cancer Res. 2006;66:3197–204.CrossRefPubMed Pore N, Jiang Z, Gupta A, Cerniglia G, Kao GD, Maity A. EGFR tyrosine kinase inhibitors decrease VEGF expression by both hypoxia-inducible factor (HIF)-1–independent and HIF-1–dependent mechanisms. Cancer Res. 2006;66:3197–204.CrossRefPubMed
31.
go back to reference Maiuri MC, Criollo A, Tasdemir E, Vicencio JM, Tajeddine N, Hickman JA, et al. BH3-Only proteins and BH3 mimetics induce autophagy by competitively disrupting the interaction between Beclin 1 and Bcl-2/Bcl-X(L). Autophagy. 2007;3:374–6.CrossRefPubMed Maiuri MC, Criollo A, Tasdemir E, Vicencio JM, Tajeddine N, Hickman JA, et al. BH3-Only proteins and BH3 mimetics induce autophagy by competitively disrupting the interaction between Beclin 1 and Bcl-2/Bcl-X(L). Autophagy. 2007;3:374–6.CrossRefPubMed
32.
go back to reference Oberstein A, Jeffrey PD, Shi Y. Crystal structure of the Bcl-XL-Beclin 1 peptide complex. J Biol Chem. 2007;282:13123–32.CrossRefPubMed Oberstein A, Jeffrey PD, Shi Y. Crystal structure of the Bcl-XL-Beclin 1 peptide complex. J Biol Chem. 2007;282:13123–32.CrossRefPubMed
33.
go back to reference Jiang ZF, Shao LJ, Wang WM, Yan XB, Liu RY. Decreased expression of Beclin-1 and LC3 in human lung cancer. Mol Biol Rep. 2012;39(1):259–67.CrossRefPubMed Jiang ZF, Shao LJ, Wang WM, Yan XB, Liu RY. Decreased expression of Beclin-1 and LC3 in human lung cancer. Mol Biol Rep. 2012;39(1):259–67.CrossRefPubMed
34.
go back to reference Shen Y, Li DD, Wang LL, Deng R, Zhu XF. Decreased expression of autophagy-related proteins in malignant epithelial ovarian cancer. Autophagy. 2008;4(8):1067–8.CrossRefPubMed Shen Y, Li DD, Wang LL, Deng R, Zhu XF. Decreased expression of autophagy-related proteins in malignant epithelial ovarian cancer. Autophagy. 2008;4(8):1067–8.CrossRefPubMed
35.
go back to reference Yoshioka A, Miyata H, Doki Y, Yamasaki M, Sohma I, Gotoh K, et al. LC3, an autophagosome marker, is highly expressed in gastrointestinal cancers. Int J Oncol. 2008;33(3):461–8.PubMed Yoshioka A, Miyata H, Doki Y, Yamasaki M, Sohma I, Gotoh K, et al. LC3, an autophagosome marker, is highly expressed in gastrointestinal cancers. Int J Oncol. 2008;33(3):461–8.PubMed
36.
go back to reference Karpathiou G, Sivridis E, Koukourakis MI, Mikroulis D, Bouros D, Froudarakis ME, et al. Light-chain 3A autophagic activity and prognostic significance in non-small cell lung carcinomas. Chest. 2011;140(1):127–34.CrossRefPubMed Karpathiou G, Sivridis E, Koukourakis MI, Mikroulis D, Bouros D, Froudarakis ME, et al. Light-chain 3A autophagic activity and prognostic significance in non-small cell lung carcinomas. Chest. 2011;140(1):127–34.CrossRefPubMed
37.
go back to reference Scanlon SE, Glazer PM. Multifaceted control of DNA repair pathways by the hypoxic tumor microenvironment. DNA Repair (Amst). 2015;32:180–9.CrossRef Scanlon SE, Glazer PM. Multifaceted control of DNA repair pathways by the hypoxic tumor microenvironment. DNA Repair (Amst). 2015;32:180–9.CrossRef
Metadata
Title
Autophagy contributes to the chemo-resistance of non-small cell lung cancer in hypoxic conditions
Authors
Jin Gu Lee
Ju Hye Shin
Hyo Sup Shim
Chang Young Lee
Dae Joon Kim
Young Sam Kim
Kyung Young Chung
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Respiratory Research / Issue 1/2015
Electronic ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-015-0285-4

Other articles of this Issue 1/2015

Respiratory Research 1/2015 Go to the issue